Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs

Tailin He , Lei Qin , Sheng Chen , Shaochuan Huo , Jie Li , Fuping Zhang , Weihong Yi , Yifang Mei , Guozhi Xiao

Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 49

PDF
Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 49 DOI: 10.1038/s41413-025-00424-1
Review Article

Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs

Author information +
History +
PDF

Abstract

Bone has long been acknowledged as a fundamental structural entity that provides support and protection to the body’s organs. However, emerging research indicates that bone plays a crucial role in the regulation of systemic metabolism. This is achieved through the secretion of a variety of hormones, cytokines, metal ions, extracellular vesicles, and other proteins/peptides, collectively referred to as bone-derived factors (BDFs). BDFs act as a medium through which bones can exert targeted regulatory functions upon various organs, thereby underscoring the profound and concrete implications of bone in human physiology. Nevertheless, there remains a pressing need for further investigations to elucidate the underlying mechanisms that inform the effects of bone on other body systems. This review aims to summarize the current findings related to the roles of these significant modulators across different organs and metabolic contexts by regulating critical genes and signaling pathways in vivo. It also addresses their involvement in the pathogenesis of various diseases affecting the musculoskeletal system, circulatory system, glucose and lipid metabolism, central nervous system, urinary system, and reproductive system. The insights gained from this review may contribute to the development of innovative therapeutic strategies through a focused approach to bone secretomes. Continued research into BDFs is expected to enhance our understanding of bone as a multifunctional organ with diverse regulatory roles in human health.

Keywords

Biological Sciences / Biochemistry and Cell Biology

Cite this article

Download citation ▾
Tailin He, Lei Qin, Sheng Chen, Shaochuan Huo, Jie Li, Fuping Zhang, Weihong Yi, Yifang Mei, Guozhi Xiao. Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs. Bone Research, 2025, 13(1): 49 DOI:10.1038/s41413-025-00424-1

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

YaoY, CaiX, ChenY, ZhangM, ZhengC. Estrogen deficiency-mediated osteoimmunity in postmenopausal osteoporosis. Med. Res. Rev., 2024, 45: 561-575.

[2]

Parfitt, A. M. The cellular basis of bone turnover and bone loss: a rebuttal of the osteocytic resorption–bone flow theory. Clin. Orthop. Relat. Res. 127, 236–247 (1977).

[3]

TresguerresFGF, et al. . The osteocyte: a multifunctional cell within the bone. Ann. Anat., 2020, 227. 151422

[4]

LiangW, et al. . An integrated multi-omics analysis reveals osteokines involved in global regulation. Cell Metab., 2024, 36: 1144-1163.e7.

[5]

DuY, ZhangL, WangZ, ZhaoX, ZouJ. Endocrine regulation of extra-skeletal organs by bone-derived secreted protein and the effect of mechanical stimulation. Front. Cell Dev. Biol., 2021, 9: 778015.

[6]

WangH, et al. . The endocrine role of bone: novel functions of bone-derived cytokines. Biochem. Pharmacol., 2021, 183: 114308.

[7]

LiY, et al. . Bone-. Front. Endocrinol., 2019, 10: 846.

[8]

BonnetN. Bone-derived factors: a new gateway to regulate glycemia. Calcif. Tissue Int., 2017, 100: 174-183.

[9]

ChenH, ShangD, WenY, LiangC. Bone-derived modulators that regulate brain function: emerging therapeutic targets for neurological disorders. Front. Cell Dev. Biol., 2021, 9: 683457.

[10]

YangM, et al. . Bone-kidney axis: a potential therapeutic target for diabetic nephropathy. Front. Endocrinol., 2022, 13. 996776

[11]

JinF, LiuM, ZhangD, WangX. Translational perspective on bone-derived cytokines in inter-organ communications. Innovation, 2023, 4: 100365

[12]

TakashiY, KawanamiD. The role of bone-derived hormones in glucose metabolism, diabetic kidney disease, and cardiovascular disorders. Int. J. Mol. Sci., 2022, 23: 2376.

[13]

KarsentyG. Osteocalcin: a multifaceted bone-derived hormone. Annu. Rev. Nutr., 2023, 43: 55-71.

[14]

DengAF, et al. . Bone-organ axes: bidirectional crosstalk. Mil. Med. Res., 2024, 11: 37

[15]

WuC, et al. . Kindlin-2 controls TGF-beta signalling and Sox9 expression to regulate chondrogenesis. Nat. Commun., 2015, 6. 7531

[16]

ChenS, et al. . Roles of focal adhesion proteins in skeleton and diseases. Acta Pharm. Sin. B, 2023, 13: 998-1013.

[17]

EastellR, SzulcP. Use of bone turnover markers in postmenopausal osteoporosis. Lancet Diabetes Endocrinol., 2017, 5: 908-923.

[18]

KikyoN. Circadian regulation of bone remodeling. Int. J. Mol. Sci., 2024, 22: 5445

[19]

BoyceBF. Advances in the regulation of osteoclasts and osteoclast functions. J. Dent. Res., 2013, 92: 860-867.

[20]

BrownJP, Don-WauchopeA, DouvilleP, AlbertC, VasikaranSD. Current use of bone turnover markers in the management of osteoporosis. Clin. Biochem., 2022, 109-110: 1-10.

[21]

VlotMC, et al. . Clinical utility of bone markers in various diseases. Bone, 2018, 114: 215-225.

[22]

AnwarA, et al. . Sensors in bone: technologies, applications, and future directions. Sensors, 2024, 24: 6172.

[23]

HuX, et al. . Irisin as an agent for protecting against osteoporosis: A review of the current mechanisms and pathways. J. Adv. Res., 2023, 62: 175-186.

[24]

FinnesTE, et al. . Procollagen type 1 amino-terminal propeptide (P1NP) and risk of hip fractures in elderly Norwegian men and women. A NOREPOS study. Bone, 2014, 64: 1-7.

[25]

ShenF, HuangX, HeG, ShiY. The emerging studies on mesenchymal progenitors in the long bone. Cell Biosci., 2023, 13: 105.

[26]

SzeligaA, et al. . Bone: a neglected endocrine organ?. J. Clin. Med., 2024, 13: 3889.

[27]

BellissimoMP, et al. . Metabolomic associations with serum bone turnover markers. Nutrients, 2020, 12: 3161.

[28]

MoseshviliE, et al. . Serum procollagen 1 amino-terminal propeptide (P1NP) in prostate cancer: pitfalls of its use as an early surrogate marker for bone metastasis. J. Med. Imaging Radiat. Oncol., 2014, 58: 497-502.

[29]

SøeK. Osteoclast fusion: physiological regulation of multinucleation through heterogeneity-potential implications for drug sensitivity. Int. J. Mol. Sci., 2020, 21: 7717.

[30]

LeiY, et al. . Identification and functional analysis of tartrate-resistant acid phosphatase type 5b (TRAP5b) in Oreochromis niloticus. Int. J. Mol. Sci., 2023, 24: 7179.

[31]

HuangM, ZhaoJ, HuangY, DaiL, ZhangX. Meta-analysis of urinary C-terminal telopeptide of type II collagen as a biomarker in osteoarthritis diagnosis. J. Orthop. Transl., 2018, 13: 50-57

[32]

ChenYN, WeiP, Yu BsJ. Higher concentration of serum C-terminal cross-linking telopeptide of type I collagen is positively related with inflammatory factors in postmenopausal women with H-type hypertension and osteoporosis. Orthop. Surg., 2019, 11: 1135-1141.

[33]

Dal PráKJ, LemosCA, OkamotoR, SoubhiaAM, PellizzerEP. Efficacy of the C-terminal telopeptide test in predicting the development of bisphosphonate-related osteonecrosis of the jaw: a systematic review. Int. J. Oral. Maxillofac. Surg., 2017, 46: 151-156.

[34]

HaeriNS, PereraS, GreenspanSL. The association of bone turnover markers with muscle function, falls, and frailty in older women in long-term care. J. Frailty. Aging, 2023, 12: 284-290.

[35]

SmithC, et al. . Higher bone remodeling biomarkers are related to a higher muscle function in older adults: effects of acute exercise. Bone, 2022, 165. 116545

[36]

HauschkaPV, LianJB, ColeDE, GundbergCM. Osteocalcin and matrix Gla protein: vitamin K-dependent proteins in bone. Physiol. Rev., 1989, 69: 990-1047.

[37]

DirckxN, MoorerMC, ClemensTL, RiddleRC. The role of osteoblasts in energy homeostasis. Nat. Rev. Endocrinol., 2019, 15: 651-665.

[38]

DucyP, et al. . Increased bone formation in osteocalcin-deficient mice. Nature, 1996, 382: 448-452.

[39]

LambertLJ, et al. . Increased trabecular bone and improved biomechanics in an osteocalcin-null rat model created by CRISPR/Cas9 technology. Dis. Model Mech., 2016, 9: 1169-1179

[40]

DiegelCR, et al. . An osteocalcin-deficient mouse strain without endocrine abnormalities. PLoS Genet., 2020, 16: e1008361.

[41]

Komori, T. Functions of osteocalcin in bone, pancreas, testis, and muscle. Int. J. Mol. Sci. 21, 7513 (2020).

[42]

KarsentyG. Update on the biology of osteocalcin. Endocr. Pract., 2017, 23: 1270-1274.

[43]

CelesteAJ, et al. . Isolation of the human gene for bone gla protein utilizing mouse and rat cDNA clones. EMBO J., 1986, 5: 1885-1890.

[44]

DesboisC, HogueDA, KarsentyG. The mouse osteocalcin gene cluster contains three genes with two separate spatial and temporal patterns of expression. J. Biol. Chem., 1994, 269: 1183-1190.

[45]

KomoriT. Regulation of proliferation, differentiation and functions of osteoblasts by Runx2. Int. J. Mol. Sci., 2019, 20: 1694.

[46]

YuanG, LiZ, LinX, LiN, XuR. New perspective of skeletal stem cells. J. Biol. Chem., 2022, 3: 280-294

[47]

SiJ, WangC, ZhangD, WangB, ZhouY. Osteopontin in bone metabolism and bone diseases. Med. Sci. Monit., 2020, 26. e919159

[48]

LeongWF, ZhouT, LimGL, LiB. Protein palmitoylation regulates osteoblast differentiation through BMP-induced osterix expression. PLoS One, 2009, 4: e4135.

[49]

CalixtoRD, et al. . Effect of the secretome of mesenchymal stem cells overexpressing BMP-9 on osteoblast differentiation and bone repair. J. Cell Physiol., 2023, 238: 2625-2637.

[50]

WangLT, ChenLR, ChenKH. Hormone-related and drug-induced osteoporosis: a cellular and molecular overview. Int. J. Mol. Sci., 2023, 24: 5814.

[51]

BarfieldWL, et al. . Eccentric muscle challenge shows osteopontin polymorphism modulation of muscle damage. Hum. Mol. Genet., 2014, 23: 4043-4050.

[52]

UaesoontrachoonK, et al. . Osteopontin and skeletal muscle myoblasts: association with muscle regeneration and regulation of myoblast function in vitro. Int. J. Biochem. Cell Biol., 2008, 40: 2303-2314.

[53]

Wasgewatte WijesingheDK, MackieEJ, PagelCN. Normal inflammation and regeneration of muscle following injury require osteopontin from both muscle and non-muscle cells. Skelet. Muscle, 2019, 9. 6

[54]

WangY, et al. . Osteopontin may improve postinjury muscle repair via matrix metalloproteinases and tgf-β activation in regular exercise. Int. J. Med. Sci., 2023, 20: 1202-1211.

[55]

LiX, et al. . Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J. Biol. Chem., 2005, 280: 19883-19887.

[56]

WangN, et al. . Role of sclerostin and dkk1 in bone remodeling in type 2 diabetic patients. Endocr. Res., 2018, 43: 29-38.

[57]

TanakaS, MatsumotoT. Sclerostin: from bench to bedside. J. Bone Miner. Metab., 2021, 39: 332-340.

[58]

WilliamsBO. LRP5: from bedside to bench to bone. Bone, 2017, 102: 26-30.

[59]

SangadalaS, et al. . Sclerostin small-molecule inhibitors promote osteogenesis by activating canonical Wnt and BMP pathways. Elife, 2023, 12. e63402

[60]

McClungMR, et al. . Romosozumab in postmenopausal women with low bone mineral density. N. Engl. J. Med., 2014, 370: 412-420.

[61]

LangdahlBL, et al. . Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial. Lancet, 2017, 390: 1585-1594.

[62]

SaagKG, et al. . Romosozumab or alendronate for fracture prevention in women with osteoporosis. N. Engl. J. Med., 2017, 377: 1417-1427.

[63]

TobiasJH. Sclerostin and cardiovascular disease. Curr. Osteoporos. Rep., 2023, 21: 519-526.

[64]

SebastianA, LootsGG. Transcriptional control of Sost in bone. Bone, 2017, 96: 76-84.

[65]

GaoH, et al. . Bone marrow adipoq+ cell population controls bone mass via sclerostin in mice. Sig. Transduct. Target. Ther., 2023, 8: 265.

[66]

CaoH, et al. . Focal adhesion protein Kindlin-2 regulates bone homeostasis in mice. Bone Res., 2020, 8: 2.

[67]

QinL, et al. . Kindlin-2 mediates mechanotransduction in bone by regulating expression of Sclerostin in osteocytes. Commun. Biol., 2021, 4: 402.

[68]

WangY, et al. . Focal adhesion proteins Pinch1 and Pinch2 regulate bone homeostasis in mice. JCI Insight, 2019, 4. e131692

[69]

MagaròMS, et al. . Identification of sclerostin as a putative new myokine involved in the muscle-to-bone crosstalk. Biomedicines, 2021, 9: 71.

[70]

MorettiA, IolasconG. Sclerostin: clinical insights in muscle-bone crosstalk. J. Int. Med. Res., 2023, 51: 3000605231193293.

[71]

AryanaI, RiniSS, SoejonoCH. Importance of sclerostin as bone-muscle mediator crosstalk. Ann. Geriatr. Med. Res., 2022, 26: 72-82.

[72]

AndersonDM, et al. . A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature, 1997, 390: 175-179.

[73]

DougallWC, et al. . RANK is essential for osteoclast and lymph node development. Genes Dev., 1999, 13: 2412-2424.

[74]

SimonetWS, et al. . Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell, 1997, 89: 309-319.

[75]

KongYY, BoyleWJ, PenningerJM. Osteoprotegerin ligand: a regulator of immune responses and bone physiology. Immunol. Today, 2000, 21: 495-502.

[76]

KongYY, et al. . OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature, 1999, 397: 315-323.

[77]

FataJE, et al. . The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell, 2000, 103: 41-50.

[78]

KimNS, et al. . Receptor activator of NF-kappaB ligand regulates the proliferation of mammary epithelial cells via Id2. Mol. Cell. Biol., 2006, 26: 1002-1013.

[79]

HuY, ChenX, WangS, JingY, SuJ. Subchondral bone microenvironment in osteoarthritis and pain. Bone Res., 2021, 9: 20.

[80]

WrightHL, McCarthyHS, MiddletonJ, MarshallMJ. RANK, RANKL and osteoprotegerin in bone biology and disease. Curr. Rev. Musculoskelet. Med, 2009, 2: 56-64.

[81]

NakashimaT, et al. . Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat. Med., 2011, 17: 1231-1234.

[82]

IkedaT, KasaiM, UtsuyamaM, HirokawaK. Determination of three isoforms of the receptor activator of nuclear factor-kappaB ligand and their differential expression in bone and thymus. Endocrinology, 2001, 142: 1419-1426.

[83]

HikitaA, et al. . Identification of an alternatively spliced variant of Ca2+-promoted Ras inactivator as a possible regulator of RANKL shedding. J. Biol. Chem., 2005, 280: 41700-41706.

[84]

LeiY, et al. . LIM domain proteins Pinch1/2 regulate chondrogenesis and bone mass in mice. Bone Res., 2020, 8: 37.

[85]

JaarahN, et al. . Differential effects of teriparatide, denosumab and zoledronate on hip structural and mechanical parameters in osteoporosis; a real-life study. J. Endocrinol. Invest., 2024, 47: 1667-1677.

[86]

HsuH, et al. . Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc. Natl. Acad. Sci. USA, 1999, 96: 3540-3545.

[87]

BucayN, et al. . osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev., 1998, 12: 1260-1268.

[88]

AmizukaN, et al. . Defective bone remodelling in osteoprotegerin-deficient mice. J. Electron. Microsc., 2003, 52: 503-513.

[89]

ChenWG, et al. . The Emerging Science of Interoception: sensing, integrating, interpreting, and regulating signals within the self. Trends Neurosci., 2021, 44: 3-16.

[90]

GaoF, et al. . Brain regulates weight bearing bone through PGE2 skeletal interoception: implication of ankle osteoarthritis and pain. Bone Res., 2024, 12: 16.

[91]

ChenH, et al. . Prostaglandin E2 mediates sensory nerve regulation of bone homeostasis. Nat. Commun., 2019, 10. 181

[92]

QiaoW, et al. . Divalent metal cations stimulate skeleton interoception for new bone formation in mouse injury models. Nat. Commun., 2022, 13. 535

[93]

LiuSZ, et al. . Prostaglandin E2/cyclooxygenase pathway in human skeletal muscle: influence of muscle fiber type and age. J. Appl. Physiol., 2016, 120: 546-551.

[94]

HoATV, et al. . Prostaglandin E2 is essential for efficacious skeletal muscle stem-cell function, augmenting regeneration and strength. Proc. Natl. Acad. Sci. USA, 2017, 114: 6675-6684.

[95]

LiY, et al. . Natural plant tissue with bioinspired nano amyloid and hydroxyapatite as green scaffolds for bone regeneration. Adv. Healthc. Mater., 2022, 11. e2102807

[96]

Hou, X. et al. Calcium phosphate-based biomaterials for bone repair. J. Funct. Biomater. 13, 187 (2022).

[97]

YuH, et al. . The anti-inflammation effect of strontium ranelate on rat chondrocytes with or without IL-1β in vitro. Exp. Ther. Med., 2022, 23: 208.

[98]

MehtaKJ. Role of iron and iron-related proteins in mesenchymal stem cells: Cellular and clinical aspects. J. Cell Physiol., 2021, 236: 7266-7289.

[99]

ZhangX, ChenQ, MaoX. Magnesium enhances osteogenesis of BMSCs by tuning osteoimmunomodulation. Biomed. Res. Int., 2019, 2019: 7908205.

[100]

HiokiT, et al. . Oncostatin M enhances osteoprotegerin synthesis but reduces macrophage colony‑stimulating factor synthesis in bFGF‑stimulated osteoblast‑like cells. Exp. Ther. Med., 2024, 27: 34.

[101]

YamashitaY, HayashiM, SaitoM, NakashimaT. Osteoblast lineage cell-derived Sema3A regulates bone homeostasis independently of androgens. Endocrinology, 2022, 163. bqac126

[102]

HayashiM, et al. . Osteoprotection by semaphorin 3A. Nature, 2012, 485: 69-74.

[103]

KenanS, et al. . Investigation of the effects of semaphorin 3A on new bone formation in a rat calvarial defect model. J. Craniomaxillofac. Surg., 2019, 47: 473-483.

[104]

ZhangN, HuaY, LiY, PanJ. Sema3A accelerates bone formation during distraction osteogenesis in mice. Connect Tissue Res., 2022, 63: 382-392.

[105]

HayashiM, et al. . Autoregulation of osteocyte Sema3A orchestrates estrogen action and counteracts bone aging. Cell Metab., 2019, 29: 627-637.e625.

[106]

GreweJM, et al. . The role of sphingosine-1-phosphate in bone remodeling and osteoporosis. Bone Res., 2022, 10: 34.

[107]

WangY, et al. . Ablation of Ephrin B2 in Col2 expressing cells delays fracture repair. Endocrinology, 2020, 161. bqaa179

[108]

Negishi-KogaT, et al. . Suppression of bone formation by osteoclastic expression of semaphorin 4D. Nat. Med., 2011, 17: 1473-1480.

[109]

RutkovskiyA, StensløkkenKO, VaageIJ. Osteoblast differentiation at a Glance. Med. Sci. Monit. Basic Res., 2016, 22: 95-106.

[110]

LeeNK, et al. . Endocrine regulation of energy metabolism by the skeleton. Cell, 2007, 130: 456-469.

[111]

PiM, WuY, QuarlesLD. GPRC6A mediates responses to osteocalcin in β-cells in vitro and pancreas in vivo. J. Bone Miner. Res., 2011, 26: 1680-1683.

[112]

WeiJ, HannaT, SudaN, KarsentyG, DucyP. Osteocalcin promotes β-cell proliferation during development and adulthood through Gprc6a. Diabetes, 2014, 63: 1021-1031.

[113]

OtaniT, et al. . Signaling pathway for adiponectin expression in adipocytes by osteocalcin. Cell. Signal., 2015, 27: 532-544.

[114]

OtaniT, et al. . Osteocalcin triggers Fas/FasL-mediated necroptosis in adipocytes via activation of p300. Cell Death Dis., 2018, 9. 1194

[115]

GuedesJAC, EstevesJV, MoraisMR, ZornTM, FuruyaDT. Osteocalcin improves insulin resistance and inflammation in obese mice: participation of white adipose tissue and bone. Bone, 2018, 115: 68-82.

[116]

LiQ, et al. . T cell factor 7 (TCF7)/TCF1 feedback controls osteocalcin signaling in brown adipocytes independent of the Wnt/β-Catenin pathway. Mol. Cell. Biol., 2018, 38: e00562-17.

[117]

ZhangM, et al. . Osteocalcin alleviates nonalcoholic fatty liver disease in mice through GPRC6A. Int. J. Endocrinol., 2021, 2021: 9178616.

[118]

MeraP, et al. . Osteocalcin signaling in myofibers is necessary and sufficient for optimum adaptation to exercise. Cell Metab., 2016, 23: 1078-1092.

[119]

MeraP, LaueK, WeiJ, BergerJM, KarsentyG. Osteocalcin is necessary and sufficient to maintain muscle mass in older mice. Mol. Metab., 2016, 5: 1042-1047.

[120]

ChowdhuryS, et al. . Muscle-derived interleukin 6 increases exercise capacity by signaling in osteoblasts. J. Clin. Invest., 2020, 130: 2888-2902.

[121]

ClinkenbeardEL, et al. . Conditional deletion of murine Fgf23: interruption of the normal skeletal responses to phosphate challenge and rescue of genetic hypophosphatemia. J. Bone Miner. Res., 2016, 31: 1247-1257.

[122]

RamonI, KleynenP, BodyJJ, KarmaliR. Fibroblast growth factor 23 and its role in phosphate homeostasis. Eur. J. Endocrinol., 2010, 162: 1-10.

[123]

HoBB, BergwitzC. FGF23 signalling and physiology. J. Mol. Endocrinol., 2021, 66: R23-r32.

[124]

ShimadaT, et al. . Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J. Clin. Invest., 2004, 113: 561-568.

[125]

HesseM, FröhlichLF, ZeitzU, LanskeB, ErbenRG. Ablation of vitamin D signaling rescues bone, mineral, and glucose homeostasis in Fgf-23 deficient mice. Matrix Biol., 2007, 26: 75-84.

[126]

SinghS, et al. . Fibroblast growth factor 23 directly targets hepatocytes to promote inflammation in chronic kidney disease. Kidney Int., 2016, 90: 985-996.

[127]

AmreinK, et al. . Sclerostin and its association with physical activity, age, gender, body composition, and bone mineral content in healthy adults. J. Clin. Endocrinol. Metab., 2012, 97: 148-154.

[128]

DanieleG, et al. . Sclerostin and insulin resistance in prediabetes: evidence of a cross talk between bone and glucose metabolism. Diabetes Care, 2015, 38: 1509-1517.

[129]

FairfieldH, et al. . The skeletal cell-derived molecule sclerostin drives bone marrow adipogenesis. J. Cell. Physiol., 2018, 233: 1156-1167.

[130]

KimSP, et al. . Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. Proc. Natl. Acad. Sci. Usa., 2017, 114: E11238-e11247.

[131]

KimSP, et al. . Bone-derived sclerostin and Wnt/β-catenin signaling regulate PDGFRα+ adipoprogenitor cell differentiation. FASEB J., 2021, 35. e21957

[132]

FulzeleK, et al. . Osteocyte-secreted wnt signaling inhibitor sclerostin contributes to beige adipogenesis in peripheral fat depots. J. Bone Miner. Res., 2017, 32: 373-384.

[133]

Riddle, R. C. Endocrine functions of sclerostin. Curr. Opin. Endocr. Metab. Res. 28, https://doi.org/10.1016/j.coemr.2022.100433 (2023).

[134]

KimSP, et al. . Lrp4 expression by adipocytes and osteoblasts differentially impacts sclerostin’s endocrine effects on body composition and glucose metabolism. J. Biol. Chem., 2019, 294: 6899-6911.

[135]

KurganN, et al. . Sclerostin influences exercise-induced adaptations in body composition and white adipose tissue morphology in male mice. J. Bone Miner. Res., 2023, 38: 541-555.

[136]

GlinkaA, et al. . Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature, 1998, 391: 357-362.

[137]

YangZ, et al. . Hepatic DKK1-driven steatosis is CD36 dependent. Life Sci. Alliance, 2023, 6: e202201665.

[138]

JaschkeNP, et al. . Dickkopf1 fuels inflammatory cytokine responses. Commun. Biol., 2022, 5: 1391.

[139]

HuangY, LiuL, LiuA. Dickkopf-1: current knowledge and related diseases. Life Sci., 2018, 209: 249-254.

[140]

KikuchiA, MatsumotoS, SadaR. Dickkopf signaling, beyond Wnt-mediated biology. Semin. Cell Dev. Biol., 2022, 125: 55-65.

[141]

HildebrandtN, et al. . Role of osteogenic Dickkopf-1 in bone remodeling and bone healing in mice with type I diabetes mellitus. Sci. Rep., 2021, 11. 1920

[142]

ColditzJ, et al. . Osteogenic Dkk1 mediates glucocorticoid-induced but not arthritis-induced bone loss. J. Bone Miner. Res., 2019, 34: 1314-1323.

[143]

ColditzJ, PickeAK, HofbauerLC, RaunerM. Contributions of Dickkopf-1 to obesity-induced bone loss and marrow adiposity. JBMR, 2020, 4: e10364

[144]

WangY, et al. . Anti-DKK1 enhances the early osteogenic differentiation of human adipose-derived stem/stromal cells. Stem Cells Dev., 2020, 29: 1007-1015.

[145]

YanQW, et al. . The adipokine lipocalin 2 is regulated by obesity and promotes insulin resistance. Diabetes, 2007, 56: 2533-2540.

[146]

JaberiSA, et al. . Lipocalin-2: structure, function, distribution and role in metabolic disorders. Biomed. Pharmacother., 2021, 142: 112002.

[147]

ChiY, et al. . Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science, 2020, 369: 276-282.

[148]

DeisJA, et al. . Adipose Lipocalin 2 overexpression protects against age-related decline in thermogenic function of adipose tissue and metabolic deterioration. Mol. Metab., 2019, 24: 18-29.

[149]

YangY, LiuJ, KousteniS. Lipocalin 2-A bone-derived anorexigenic and β-cell promoting signal: From mice to humans. J. Diabetes, 2023, 16. e13504

[150]

CostaD, et al. . Altered bone development and turnover in transgenic mice over-expressing lipocalin-2 in bone. J. Cell. Physiol., 2013, 228: 2210-2221.

[151]

CapulliM, et al. . A complex role for lipocalin 2 in bone metabolism: global ablation in mice induces osteopenia caused by an altered energy metabolism. J. Bone Miner. Res., 2018, 33: 1141-1153.

[152]

MosialouI, et al. . MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Nature, 2017, 543: 385-390.

[153]

PetropoulouPI, et al. . Lipocalin-2 is an anorexigenic signal in primates. Elife, 2020, 9. e58949

[154]

MosialouI, et al. . Lipocalin-2 counteracts metabolic dysregulation in obesity and diabetes. J. Exp. Med., 2020, 217. e20191261

[155]

KidoS, et al. . Mechanical stress induces Interleukin-11 expression to stimulate osteoblast differentiation. Bone, 2009, 45: 1125-1132.

[156]

DongB, et al. . Osteoblast/osteocyte-derived interleukin-11 regulates osteogenesis and systemic adipogenesis. Nat. Commun., 2022, 13. 7194

[157]

ZhangJ, et al. . Exercise-induced irisin in bone and systemic irisin administration reveal new regulatory mechanisms of bone metabolism. Bone Res., 2017, 5: 16056.

[158]

HenneickeH, et al. . Skeletal glucocorticoid signalling determines leptin resistance and obesity in aging mice. Mol. Metab., 2020, 42: 101098.

[159]

XuR, et al. . Targeting skeletal endothelium to ameliorate bone loss. Nat. Med., 2018, 24: 823-833.

[160]

LiZ, et al. . Bone controls browning of white adipose tissue and protects from diet-induced obesity through Schnurri-3-regulated SLIT2 secretion. Nat. Commun., 2024, 15. 6697

[161]

SongZ, et al. . Osteopontin takes center stage in chronic liver disease. Hepatology, 2021, 73: 1594-1608.

[162]

DaiB, et al. . Macrophages in epididymal adipose tissue secrete osteopontin to regulate bone homeostasis. Nat. Commun., 2022, 13. 427

[163]

KatagiriT, WatabeT. Bone morphogenetic proteins. Cold Spring Harb. Perspect. Biol., 2016, 8. a021899

[164]

BabootaRK, et al. . BMP4 and Gremlin 1 regulate hepatic cell senescence during clinical progression of NAFLD/NASH. Nat. Metab., 2022, 4: 1007-1021.

[165]

SunQJ, et al. . The role of bone morphogenetic protein 9 in nonalcoholic fatty liver disease in mice. Front. Pharmacol., 2020, 11: 605967.

[166]

TrangmarSJ, González-AlonsoJ. Heat, Hydration and the Human Brain, Heart and Skeletal Muscles. Sports Med, 2019, 49: 69-85.

[167]

OttoE, et al. . Crosstalk of brain and bone-clinical observations and their molecular bases. Int. J. Mol. Sci., 2020, 21: 4946.

[168]

PiM, NishimotoSK, Darryl QuarlesL. Explaining divergent observations regarding osteocalcin/GPRC6A endocrine signaling. Endocrinology, 2021, 162: bqab011.

[169]

HouYF, et al. . Gut microbiota-derived propionate mediates the neuroprotective effect of osteocalcin in a mouse model of Parkinson’s disease. Microbiome, 2021, 9. 34

[170]

KhrimianL, et al. . Gpr158 mediates osteocalcin’s regulation of cognition. J. Exp. Med, 2017, 214: 2859-2873.

[171]

QianZ, et al. . Osteocalcin attenuates oligodendrocyte differentiation and myelination via GPR37 signaling in the mouse brain. Sci. Adv., 2021, 7: eabi5811.

[172]

VilledaSA, et al. . Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med., 2014, 20: 659-663.

[173]

OuryF, et al. . Maternal and offspring pools of osteocalcin influence brain development and functions. Cell, 2013, 155: 228-241.

[174]

GuoXZ, et al. . Osteocalcin ameliorates motor dysfunction in a 6-Hydroxydopamine-induced Parkinson’s disease rat model through AKT/GSK3β signaling. Front. Mol. Neurosci., 2018, 11: 343.

[175]

WuJ, DouY, LiuW, ZhaoY, LiuX. Osteocalcin improves outcome after acute ischemic stroke. Aging, 2020, 12: 387-396.

[176]

BhusalA, LeeWH, SukK. Lipocalin-2 in diabetic complications of the nervous system: physiology, pathology, and beyond. Front. Physiol., 2021, 12: 638112.

[177]

ZhaoRY, et al. . Role of lipocalin 2 in stroke. Neurobiol. Dis., 2023, 179: 106044.

[178]

KimBW, et al. . Pathogenic upregulation of glial lipocalin-2 in the parkinsonian dopaminergic system. J. Neurosci., 2016, 36: 5608-5622.

[179]

WangG, et al. . Neutralization of lipocalin-2 diminishes stroke-reperfusion injury. Int. J. Mol. Sci., 2020, 21: 6253.

[180]

ZhangJ, NovakovicN, HuaY, KeepRF, XiG. Role of lipocalin-2 in extracellular peroxiredoxin 2-induced brain swelling, inflammation and neuronal death. Exp. Neurol., 2021, 335: 113521.

[181]

MuchaM, et al. . Lipocalin-2 controls neuronal excitability and anxiety by regulating dendritic spine formation and maturation. Proc. Natl. Acad. Sci. USA., 2011, 108: 18436-18441.

[182]

ZhouY, et al. . Osteopontin as a candidate of therapeutic application for the acute brain injury. J. Cell. Mol. Med., 2020, 24: 8918-8929.

[183]

YimA, SmithC, BrownAM. Osteopontin/secreted phosphoprotein-1 harnesses glial-, immune-, and neuronal cell ligand-receptor interactions to sense and regulate acute and chronic neuroinflammation. Immunol. Rev., 2022, 311: 224-233.

[184]

RentsendorjA, et al. . A novel role for osteopontin in macrophage-mediated amyloid-β clearance in Alzheimer’s models. Brain Behav. Immun., 2018, 67: 163-180.

[185]

SunCM, et al. . Osteopontin attenuates early brain injury through regulating autophagy-apoptosis interaction after subarachnoid hemorrhage in rats. CNS Neurosci. Ther., 2019, 25: 1162-1172.

[186]

RogallR, et al. . Bioluminescence imaging visualizes osteopontin-induced neurogenesis and neuroblast migration in the mouse brain after stroke. Stem Cell Res. Ther., 2018, 9: 182.

[187]

HanH, et al. . Macrophage-derived osteopontin (SPP1) protects from nonalcoholic steatohepatitis. Gastroenterology, 2023, 165: 201-217.

[188]

Melero-JerezC, et al. . Myeloid-derived suppressor cells support remyelination in a murine model of multiple sclerosis by promoting oligodendrocyte precursor cell survival, proliferation, and differentiation. Glia, 2021, 69: 905-924.

[189]

WangJ, et al. . Osteopontin potentiates PM-induced IL-1α and IL-1β production via the ERK/JNK signaling pathway. Ecotoxicol. Environ. Saf., 2019, 171: 467-474.

[190]

CulibrkRA, ArabiyatAS, DeKalbCA, HahnMS. Modeling sympathetic hyperactivity in Alzheimer’s related bone loss. J. Alzheimers Dis., 2021, 84: 647-658.

[191]

KlementJD, et al. . An osteopontin/CD44 immune checkpoint controls CD8+ T cell activation and tumor immune evasion. J. Clin. Invest., 2018, 128: 5549-5560.

[192]

HanadaR, et al. . Central control of fever and female body temperature by RANKL/RANK. Nature, 2009, 462: 505-509.

[193]

GuerriniMM, et al. . Human osteoclast-poor osteopetrosis with hypogammaglobulinemia due to TNFRSF11A (RANK) mutations. Am. J. Hum. Genet., 2008, 83: 64-76.

[194]

GuerriniMM, et al. . Inhibition of the TNF family cytokine RANKL prevents autoimmune inflammation in the central nervous system. Immunity, 2015, 43: 1174-1185.

[195]

ShimamuraM, et al. . OPG/RANKL/RANK axis is a critical inflammatory signaling system in ischemic brain in mice. Proc. Natl. Acad. Sci. Usa., 2014, 111: 8191-8196.

[196]

WangJ, et al. . Bone-derived PDGF-BB drives brain vascular calcification in male mice. J. Clin. Invest., 2023, 133. e168447

[197]

SanthanamL, et al. . Skeleton-secreted PDGF-BB mediates arterial stiffening. J. Clin. Invest., 2021, 131. e147116

[198]

MaoY, et al. . Fibroblast growth factor-2/platelet-derived growth factor enhances atherosclerotic plaque stability. J. Cell. Mol. Med., 2020, 24: 1128-1140.

[199]

HuangH, KongL, LuanS, QiC, WuF. Ligustrazine suppresses platelet-derived growth factor-BB-induced pulmonary artery smooth muscle cell proliferation and inflammation by regulating the PI3K/AKT signaling pathway. Am. J. Chin. Med., 2021, 49: 437-459.

[200]

ZhaoZ, ZhangG, YangJ, LuR, HuH. DLEU2 modulates proliferation, migration and invasion of platelet-derived growth factor-BB (PDGF-BB)-induced vascular smooth muscle cells (VSMCs) via miR-212-5p/YWHAZ axis. Cell Cycle, 2022, 21: 2013-2026.

[201]

WangM, et al. . Circ_CHFR promotes platelet-derived growth factor-bb-induced proliferation, invasion, and migration in vascular smooth muscle cells via the miR-149-5p/NRP2 Axis. J. Cardiovasc. Pharmacol., 2022, 79: e94-e102.

[202]

FanK, et al. . Circ_0004872 promotes platelet-derived growth factor-BB-induced proliferation, migration and dedifferentiation in HA-VSMCs via miR-513a-5p/TXNIP axis. Vasc. Pharmacol., 2021, 140: 106842.

[203]

DutkaM, et al. . Osteoprotegerin and RANKL-RANK-OPG-TRAIL signalling axis in heart failure and other cardiovascular diseases. Heart Fail. Rev., 2022, 27: 1395-1411.

[204]

BennettBJ, et al. . Osteoprotegerin inactivation accelerates advanced atherosclerotic lesion progression and calcification in older ApoE−/− mice. Arterioscler Thromb. Vasc. Biol., 2006, 26: 2117-2124.

[205]

DawsonS, LawrieA. From bones to blood pressure, developing novel biologic approaches targeting the osteoprotegein pathway for pulmonary vascular disease. Pharmacol. Ther., 2017, 169: 78-82.

[206]

RochetteL, et al. . The role of osteoprotegerin and its ligands in vascular function. Int. J. Mol. Sci., 2019, 20: 705.

[207]

VergaroG, et al. . Discharge FGF23 level predicts one year outcome in patients admitted with acute heart failure. Int. J. Cardiol., 2021, 336: 98-104.

[208]

PoelzlG, et al. . FGF23 is associated with disease severity and prognosis in chronic heart failure. Eur. J. Clin. Invest., 2014, 44: 1150-1158.

[209]

EdmonstonD, GrabnerA, WolfM. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat. Rev. Cardiol., 2024, 21: 11-24.

[210]

JiaZ, et al. . Klotho/FGF23 axis regulates cardiomyocyte apoptosis and cytokine release through ERK/MAPK pathway. Cardiovasc. Toxicol., 2023, 23: 317-328.

[211]

LeidnerAS, et al. . Fibroblast growth factor 23 and risk of heart failure subtype: the CRIC (Chronic Renal Insufficiency Cohort) Study. Kidney Med., 2023, 5: 100723.

[212]

Garcia-de Los RíosC, et al. . Sclerostin as a biomarker of cardiovascular risk in women with systemic lupus erythematosus. Sci. Rep., 2022, 12. 21621

[213]

YuY, et al. . Targeting loop3 of sclerostin preserves its cardiovascular protective action and promotes bone formation. Nat. Commun., 2022, 13. 4241

[214]

LangdahlBL, HofbauerLC, ForfarJC. Cardiovascular safety and sclerostin inhibition. J. Clin. Endocrinol. Metab., 2021, 106: 1845-1853.

[215]

GolledgeJ, ThanigaimaniS. Role of sclerostin in cardiovascular disease. Arterioscler. Thromb. Vasc. Biol., 2022, 42: e187-e202.

[216]

JohnstonRA, et al. . Morphological and genomic shifts in mole-rat ‘queens’ increase fecundity but reduce skeletal integrity. Elife, 2021, 10. e65760

[217]

SchwetzV, et al. . Osteocalcin is not a strong determinant of serum testosterone and sperm count in men from infertile couples. Andrology, 2013, 1: 590-594.

[218]

KanazawaI, et al. . Undercarboxylated osteocalcin is positively associated with free testosterone in male patients with type 2 diabetes mellitus. Osteoporos. Int., 2013, 24: 1115-1119.

[219]

YaghobinejadM, et al. . Osteocalcin improves testicular morphology but does not ameliorate testosterone synthesis signaling in azoospermic mice. Clin. Exp. Reprod. Med., 2024, 51: 344-352.

[220]

SakiF, KasaeeSR, SadeghianF, KoohpeymaF, OmraniGHR. Investigating the effect of testosterone by itself and in combination with letrozole on 1,25-dihydroxy vitamin D and FGF23 in male rats. J. Endocrinol. Invest., 2019, 42: 19-25.

[221]

ZhuZ, et al. . Pregranulosa cell-derived FGF23 protects oocytes from premature apoptosis during primordial follicle formation by inhibiting p38 MAPK in mice. J. Biol. Chem., 2023, 299: 104776.

[222]

StenhouseC, et al. . Novel mineral regulatory pathways in ovine pregnancy: I. phosphate, klotho signaling, and sodium-dependent phosphate transporters. Biol. Reprod., 2021, 104: 1084-1096.

[223]

KarsentyG, OuryF. Regulation of male fertility by the bone-derived hormone osteocalcin. Mol. Cell. Endocrinol., 2014, 382: 521-526.

[224]

OuryF, et al. . Osteocalcin regulates murine and human fertility through a pancreas-bone-testis axis. J. Clin. Invest., 2013, 123: 2421-2433.

[225]

ParavatiR, et al. . Differential regulation of osteopontin and CD44 correlates with infertility status in PCOS patients. J. Mol. Med. (Berl.), 2020, 98: 1713-1725.

[226]

HeZ, et al. . Osteopontin and integrin αvβ3 expression during the implantation window in IVF patients with elevated serum progesterone and oestradiol level. Geburtshilfe Frauenheilkd., 2016, 76: 709-717.

[227]

GérardN, RobinE. Cellular and molecular mechanisms of the preovulatory follicle differenciation and ovulation: what do we know in the mare relative to other species. Theriogenology, 2019, 130: 163-176.

[228]

TríbuloP, et al. . Consequences of exposure of embryos produced in vitro in a serum-containing medium to dickkopf-related protein 1 and colony stimulating factor 2 on blastocyst yield, pregnancy rate, and birth weight. J. Anim. Sci., 2017, 95: 4407-4412.

[229]

LiuY, et al. . Excessive ovarian stimulation up-regulates the Wnt-signaling molecule DKK1 in human endometrium and may affect implantation: an in vitro co-culture study. Hum. Reprod., 2010, 25: 479-490.

[230]

ZaidiS, et al. . Regulation of FSH receptor promoter activation in the osteoclast. Biochem. Biophys. Res. Commun., 2007, 361: 910-915.

[231]

Juel MortensenL, et al. . Possible link between FSH and RANKL release from adipocytes in men with impaired gonadal function including Klinefelter syndrome. Bone, 2019, 123: 103-114.

[232]

BärL, StournarasC, LangF, FöllerM. Regulation of fibroblast growth factor 23 (FGF23) in health and disease. FEBS Lett., 2019, 593: 1879-1900.

[233]

GattineniJ, et al. . Regulation of renal phosphate transport by FGF23 is mediated by FGFR1 and FGFR4. Am. J. Physiol. Ren. Physiol., 2014, 306: F351-F358.

[234]

UrakawaI, et al. . Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature, 2006, 444: 770-774.

[235]

ChenG, et al. . α-Klotho is a non-enzymatic molecular scaffold for FGF23 hormone signalling. Nature, 2018, 553: 461-466.

[236]

LimK, et al. . α-Klotho expression in human tissues. J. Clin. Endocrinol. Metab., 2015, 100: E1308-E1318.

[237]

ShimadaT, et al. . Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc. Natl. Acad. Sci. Usa., 2001, 98: 6500-6505.

[238]

ShimadaT, et al. . FGF-23 transgenic mice demonstrate hypophosphatemic rickets with reduced expression of sodium phosphate cotransporter type IIa. Biochem. Biophys. Res. Commun., 2004, 314: 409-414.

[239]

ShimadaT, et al. . FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J. Bone Miner. Res., 2004, 19: 429-435.

[240]

LiuS, VierthalerL, TangW, ZhouJ, QuarlesLD. FGFR3 and FGFR4 do not mediate renal effects of FGF23. J. Am. Soc. Nephrol., 2008, 19: 2342-2350.

[241]

AndrukhovaO, et al. . FGF23 regulates renal sodium handling and blood pressure. EMBO Mol. Med., 2014, 6: 744-759.

[242]

GattineniJ, et al. . FGF23 decreases renal NaPi-2a and NaPi-2c expression and induces hypophosphatemia in vivo predominantly via FGF receptor 1. Am. J. Physiol. Ren. Physiol., 2009, 297: F282-F291.

[243]

AndrukhovaO, et al. . FGF23 acts directly on renal proximal tubules to induce phosphaturia through activation of the ERK1/2–SGK1 signaling pathway. Bone, 2012, 51: 621-628.

[244]

MorenaM, et al. . Osteoprotegerin and sclerostin in chronic kidney disease prior to dialysis: potential partners in vascular calcifications. Nephrol. Dial. Transplant., 2015, 30: 1345-1356.

[245]

BernardiS, et al. . Circulating osteoprotegerin is associated with chronic kidney disease in hypertensive patients. BMC Nephrol., 2017, 18: 219.

[246]

ChangYH, et al. . Serum osteoprotegerin and tumor necrosis factor related apoptosis inducing-ligand (TRAIL) are elevated in type 2 diabetic patients with albuminuria and serum osteoprotegerin is independently associated with the severity of diabetic nephropathy. Metabolism, 2011, 60: 1064-1069.

[247]

MarquesGL, et al. . Osteoprotegerin is a marker of cardiovascular mortality in patients with chronic kidney disease stages 3–5. Sci. Rep., 2021, 11. 2473

[248]

KamińskaJ, et al. . Circulating osteoprotegerin in chronic kidney disease and all-cause mortality. Int. J. Gen. Med., 2021, 14: 2413-2420.

[249]

KaeslerN, et al. . Sclerostin deficiency modifies the development of CKD-MBD in mice. Bone, 2018, 107: 115-123.

[250]

SabbaghY, et al. . Repression of osteocyte Wnt/β-catenin signaling is an early event in the progression of renal osteodystrophy. J. Bone Miner. Res., 2012, 27: 1757-1772.

[251]

Hsu, Y. C. et al. Dickkopf-1 acts as a profibrotic mediator in progressive chronic kidney disease. Int. J. Mol. Sci. 24, 7679 (2023).

[252]

LinCL, et al. . Dickkopf-1 promotes hyperglycemia-induced accumulation of mesangial matrix and renal dysfunction. J. Am. Soc. Nephrol., 2010, 21: 124-135.

[253]

CourbonG, et al. . Lipocalin 2 stimulates bone fibroblast growth factor 23 production in chronic kidney disease. Bone Res., 2021, 9: 35.

[254]

AsafS, et al. . Lipocalin 2—not only a biomarker: a study of current literature and systematic findings of ongoing clinical trials. Immunol. Res., 2023, 71: 287-313.

[255]

TsukasakiM, TakayanagiH. Osteoimmunology: evolving concepts in bone-immune interactions in health and disease. Nat. Rev. Immunol., 2019, 19: 626-642.

[256]

KogaT, et al. . Costimulatory signals mediated by the ITAM motif cooperate with RANKL for bone homeostasis. Nature, 2004, 428: 758-763.

[257]

OmatsuY. Cellular niches for hematopoietic stem cells in bone marrow under normal and malignant conditions. Inflamm. Regen., 2023, 43: 15.

[258]

HeT, et al. . The bone-liver interaction modulates immune and hematopoietic function through Pinch-Cxcl12-Mbl2 pathway. Cell Death Differ., 2024, 31: 90-105.

[259]

OmatsuY, et al. . The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity, 2010, 33: 387-399.

[260]

KolletO, et al. . Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat. Med., 2006, 12: 657-664.

[261]

FrodermannV, et al. . Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nat. Med., 2019, 25: 1761-1771.

[262]

YuVW, et al. . Specific bone cells produce DLL4 to generate thymus-seeding progenitors from bone marrow. J. Exp. Med., 2015, 212: 759-774.

[263]

TerashimaA, et al. . Sepsis-induced osteoblast ablation causes immunodeficiency. Immunity, 2016, 44: 1434-1443.

[264]

IshikawaM, et al. . Bone marrow plasma cells require P2RX4 to sense extracellular ATP. Nature, 2024, 626: 1102-1107.

[265]

KodeA, et al. . Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts. Nature, 2014, 506: 240-244.

[266]

DongL, et al. . Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment. Nature, 2016, 539: 304-308.

[267]

D’AmicoL, et al. . Dickkopf-related protein 1 (Dkk1) regulates the accumulation and function of myeloid derived suppressor cells in cancer. J. Exp. Med., 2016, 213: 827-840.

[268]

RaaijmakersMH, et al. . Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature, 2010, 464: 852-857.

[269]

CasimiroS, VilhaisG, GomesI, CostaL. The roadmap of RANKL/RANK pathway in cancer. Cells, 2021, 10: 1978.

[270]

Föger-SamwaldU, DovjakP, Azizi-SemradU, Kerschan-SchindlK, PietschmannP. Osteoporosis: pathophysiology and therapeutic options. EXCLI J., 2020, 19: 1017-1037

[271]

CadieuxB, et al. . Experience with denosumab (XGEVA®) for prevention of skeletal-related events in the 10 years after approval. J. Bone Oncol., 2022, 33: 100416.

[272]

VoggB, et al. . The totality of evidence for SDZ-deno: a biosimilar to reference denosumab. Clin. Ther., 2024, 46: 916-926.

[273]

GaleaGL, LanyonLE, PriceJS. Sclerostin’s role in bone’s adaptive response to mechanical loading. Bone, 2017, 96: 38-44.

[274]

TsourdiE, RachnerTD, HofbauerLC. Romosozumab versus alendronate and fracture risk in women with osteoporosis. N. Engl. J. Med., 2018, 378: 195

[275]

Movérare-SkrticS, et al. . B4GALNT3 regulates glycosylation of sclerostin and bone mass. EBioMedicine, 2023, 91. 104546

[276]

ArendR, et al. . DKK1 is a predictive biomarker for response to DKN-01: results of a phase 2 basket study in women with recurrent endometrial carcinoma. Gynecol. Oncol., 2023, 172: 82-91.

[277]

KatohM, KatohM. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int. J. Mol. Med., 2017, 40: 587-606

[278]

YangX, QiY, WangS. DKN-01 suppresses gastric cancer progression through activating cGAS-STING pathway to block macrophage M2 polarization. Appl. Biochem. Biotechnol., 2025, 197: 1025-1038.

[279]

Wise, D. R. et al. A Phase 1/2 multicenter trial of DKN-01 as monotherapy or in combination with docetaxel for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Prostate Cancer Prostatic Dis. https://doi.org/10.1038/s41391-024-00798-z (2024). Online ahead of print.

[280]

WallJA, KlempnerSJ, ArendRC. The anti-DKK1 antibody DKN-01 as an immunomodulatory combination partner for the treatment of cancer. Expert Opin. Investig. Drugs, 2020, 29: 639-644.

[281]

OuryF, et al. . Osteocalcin regulates murine and human fertility through a pancreas-bone-testis axis. J. Clin. Invest., 2015, 125: 2180.

Funding

National Natural Science Foundation of China (National Science Foundation of China)(82430078, 82261160395, 82230081)

the SMART Grant (B2402033), the Shenzhen Fundamental Research Program (JCYJ20220818100617036), the National Key Research and Development Program of China Grants (2019YFA0906004), the Guangdong Provincial Science and Technology Innovation Council Grant (2017B030301018), and the Shenzhen Key Laboratory of Cell Microenvironment Grant (ZDSYS20140509142721429)

Shenzhen Medical Research Funds (C2401029)

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

303

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/